Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Osteoporos Int ; 26(3): 987-95, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25391248

RESUMEN

UNLABELLED: We demonstrate histological evidence for hyperparathyroidism in patients with gastrectomy. This is, at least in part, explained by impaired calcium absorption, resulting in mineralization defects and secondary hyperparathyroidism. Additionally, we demonstrate improved bone mineralization in patients with gastrectomy after gluconate therapy and showed the effectiveness of calcium gluconate over carbonate to balance impaired calcium hemostasis in mice. INTRODUCTION: Gastrectomy and hypochlorhydria due to long-term proton pump inhibitor therapy are associated with increased fracture risk because of intestinal calcium malabsorption. Hence, our objectives were to histologically investigate bone metabolism in patients with gastrectomy and to analyze the impact of calcium gluconate supplementation on skeletal integrity in the setting of impaired gastric acidification. METHODS: Undecalcified bone biopsies of 26 gastrectomized individuals were histologically analyzed. In the clinical setting, we retrospectively identified 5 gastrectomized patients with sufficient vitamin D level, who were additionally supplemented with calcium gluconate and had a real bone mineral density (aBMD) follow-up assessments. A mouse model of achlorhydria (ATP4b-/-) was used to compare the effect of calcium gluconate and calcium carbonate supplementation on bone metabolism. RESULTS: Biopsies from gastrectomized individuals showed significantly increased osteoid, osteoclast, and osteoblast indices and fibroosteoclasia (p < 0.05) as well as impaired calcium distribution in mineralized bone matrix compared to healthy controls. Five gastrectomized patients with sufficient vitamin D level demonstrated a significant increase in aBMD after a treatment with calcium gluconate alone for at least 6 months (p < 0.05). Calcium gluconate was superior to calcium carbonate in maintaining calcium metabolism in a mouse model of achlorhydria. CONCLUSION: Gastrectomy is associated with severe osteomalacia, marrow fibrosis, and impaired calcium distribution within the mineralized matrix. We show that calcium gluconate supplementation can increase bone mineral density in gastrectomized individuals and performs superior to calcium carbonate in restoring calcium/skeletal homoeostasis in a mouse model of achlorhydria.


Asunto(s)
Gluconato de Calcio/uso terapéutico , Gastrectomía/efectos adversos , Hiperparatiroidismo Secundario/tratamiento farmacológico , Osteoporosis/tratamiento farmacológico , Aclorhidria/tratamiento farmacológico , Anciano , Animales , Biopsia , Densidad Ósea/efectos de los fármacos , Calcio/metabolismo , Gluconato de Calcio/farmacología , Carbamatos/uso terapéutico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Femenino , Homeostasis/efectos de los fármacos , Humanos , Hiperparatiroidismo Secundario/etiología , Hiperparatiroidismo Secundario/metabolismo , Ilion/patología , Masculino , Ratones Noqueados , Persona de Mediana Edad , Osteoblastos/patología , Osteoclastos/patología , Osteoporosis/etiología , Osteoporosis/patología , Osteoporosis/fisiopatología , Estudios Retrospectivos
3.
J Pathol ; 207(1): 43-52, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15983982

RESUMEN

H+/K+-ATPase beta-subunit-deficient mice (129/Sv background) display numerous pathologies in the stomach. Expression of the mutation in BALB/cCrSlc mice results in the development of an aberrant 'mucus-rich' cell population. 'Mucus-rich' cells have been described in stomachs of mice with autoimmune gastritis, a disease mediated by CD4+ T cells. Other pathological features of autoimmune gastritis are similar to those in H+/K+ beta-deficient mice and include a mononuclear cell infiltrate in the gastric mucosa, non-functional or absent parietal cells, depletion of zymogenic cells, hypergastrinaemia, and gastric unit hypertrophy caused by immature cell hyperplasia. The present study investigates further the aberrant gastric 'mucus-rich' cell lineage and analyses the mRNA expression of mucus cell products TFF1 and TFF2. 'Mucus-rich' cells stained for both acidic and neutral mucins, and with a TFF2-specific antibody. Stomachs from both models expressed decreased TFF1 mRNA and reciprocally increased TFF2 mRNA. The involvement of gastrin in regulating trefoil mRNA expression was also investigated using gastrin-deficient mice. In contrast to previous findings, gastrin did not positively regulate TFF1 mRNA expression, but there was possible augmentation of TFF2. Additionally, a clear role for inflammation was established involving both polymorphonuclear and mononuclear cells in these models, and a link was found between mucosal hypertrophy and increased interleukin-11 (IL-11) expression.


Asunto(s)
Mucosa Gástrica/patología , Gastritis/metabolismo , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Péptidos/metabolismo , Animales , Enfermedades Autoinmunes/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Citocinas/fisiología , Modelos Animales de Enfermedad , Mucosa Gástrica/metabolismo , Gastrinas/sangre , Regulación de la Expresión Génica , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Hiperplasia/metabolismo , Hipertrofia/metabolismo , Interleucina-11/biosíntesis , Interleucina-11/genética , Interleucina-11/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Mucinas/genética , Mucinas/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Péptidos/genética , Péptidos/fisiología , ARN Mensajero/genética , Especificidad de la Especie , Factor Trefoil-1 , Factor Trefoil-2
4.
Am J Physiol Gastrointest Liver Physiol ; 281(6): G1502-11, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11705756

RESUMEN

The gastric H(+)/K(+)-ATPase is essential for normal development of parietal cells. Here we have directly assessed the role of the H(+)/K(+)-ATPase beta-subunit (H/K-beta) on epithelial cell development by detailed quantitation of the epithelial cell types of the gastric mucosa of H/K-beta-deficient mice. H/K-beta-deficient mice had a 3.1-fold increase in the number of immature cells per gastric unit; however, the numbers of surface mucous and parietal cells were similar to those in the gastric units of wild-type mice. The effect of elevated gastrin levels in the H/K-beta-deficient mice was determined by producing mice that are also deficient in gastrin. We demonstrated that the increased production of immature cells and resulting hypertrophy is caused by the overproduction of gastrin. However, the depletion of zymogenic cells, which is another feature of H/K-beta-deficient mice, is independent of hypergastrinemia. Significantly, parietal cells of H/K-beta- and gastrin-deficient mice had abnormal secretory membranes and were devoid of resting tubulovesicular membranes. Together these data suggest a homeostatic mechanism limiting the number of immature cells that can develop into end-stage epithelial cells and indicate a direct role for H/K-beta in the development of mature parietal cells.


Asunto(s)
Mucosa Gástrica/patología , Gastrinas/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , Animales , Recuento de Células , Muerte Celular , División Celular , Ciclinas/análisis , Células Epiteliales/patología , Gastrinas/genética , Gastrinas/fisiología , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/fisiología , Concentración de Iones de Hidrógeno , Hipertrofia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Electrónica , Células Parietales Gástricas/patología , Fenotipo , Antígeno Nuclear de Célula en Proliferación/análisis
5.
Immunology ; 104(4): 392-401, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11899424

RESUMEN

Autoimmune gastritis, in which the H+/K(+)-ATPase of parietal cells is the major antigen, is one of the most common autoimmune diseases. Here we examined if specific properties of the H+/K(+)-ATPase or parietal cells are involved in rendering them autoimmune targets. The model antigens beta-galactosidase and ovalbumin (OVA) were expressed in parietal cells of transgenic mice. On experimental induction of autoimmune gastritis by neonatal thymectomy, autoantibodies to beta-galactosidase developed in mice expressing beta-galactosidase in parietal cells, a response that was independent of either the response to the gastric H+/K(+)-ATPase or gastric inflammation. In contrast, mice that expressed OVA in parietal cells did not exhibit an antibody response to OVA after thymectomy. However, increasing the frequency of anti-OVA T lymphocytes in OVA-expressing mice resulted in autoantibodies to OVA and gastritis. These studies indicate that parietal cells can present a variety of antigens to the immune system. Factors such as the identity and expression level of the autoantigen and the frequency of autoreactive T cells play a role in determining the prevalence and outcome of the particular immune response. In addition, as not all mice of a particular genotype displayed autoimmunity, random events are involved in determining the target of autoimmune recognition.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Gastritis/inmunología , Estómago/inmunología , Animales , Enfermedades Autoinmunes/patología , Autoinmunidad , Femenino , Gastritis/patología , Expresión Génica/inmunología , ATPasa Intercambiadora de Hidrógeno-Potásio/inmunología , Tolerancia Inmunológica , Inmunoglobulina G/biosíntesis , Masculino , Ratones , Ratones Transgénicos , Ovalbúmina/metabolismo , Células Parietales Gástricas/inmunología , Timo/inmunología , Transgenes , beta-Galactosidasa/metabolismo
6.
Autoimmunity ; 34(2): 147-54, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11905844

RESUMEN

A number of experimental models of organ-specific autoimmunity involve a period of peripheral T cell lymphopenia prior to disease onset. In particular, experimental autoimmune gastritis, induced in susceptible mouse strains by neonatal thymectomy, is a CD4+ T cell mediated autoimmune disease. We have previously demonstrated that this disease displays the hallmarks of a Th1-mediated DTH inflammatory response with an essential role for IFN-gamma very early in the pathogenesis of disease. Given the interplay between the innate and adaptive immune responses, a potential source of early IFN-gamma production in these lymphopenic mice is the innate immune response. Here we have assessed the contribution of innate immunity to the induction of experimental autoimmune gastritis, in particular, the role of natural killer (NK) cells in production of IFN-gamma. Analysis of NK cells and macrophages revealed no difference in either the number or activation status between euthymic and neonatally thymectomised mice. Furthermore, in vivo depletion of NK cells immediately after neonatal thymectomy of (BALB/cCrSlcxC57BL/6) F1 mice demonstrated no reduction in disease incidence compared to control groups of neonatally thymectomised mice. Therefore, we conclude that NK cells are not the primary source of IFN-gamma required for the pathogenesis of autoimmune gastritis following neonatal thymectomy but rather the small cohort of T cells in the periphery of lymphopenic mice are likely to be responsible for the IFN-gamma production.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Gastritis/inmunología , Células Asesinas Naturales/inmunología , Animales , Enfermedades Autoinmunes/etiología , Modelos Animales de Enfermedad , Gastritis/etiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Timectomía , Timo/cirugía
7.
Immunogenetics ; 53(9): 741-50, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11862406

RESUMEN

BALB/c mice thymectomized on their third day of life develop a high incidence of experimental autoimmune gastritis (EAG) which closely resembles human chronic atrophic (type A, autoimmune) gastritis. Linkage analysis of (BALB/cCrSlcxC57BL/6)F2 mice previously demonstrated that the Gasa1 and Gasa2 genes on distal Chromosome (Chr) 4 have major effects on the development of EAG in this murine model, while other loci displayed a trend towards linkage. Here, we implemented partitioned chi(2)-analysis in order to develop a better understanding of the genotypes contributing to susceptibility and resistance at each linkage region. This approach revealed that linkage of Gasa1 and Gasa2 to EAG was due to codominant and recessive BALB/cCrSlc alleles, respectively. To identify additional EAG susceptibility genes, separate linkage studies were performed on Gasa1 heterozygotes and Gasa2 C57BL/6 homozygotes plus heterozygotes so as to minimize the effects of these disease genes. The enhanced sensitivity of these analyses confirmed the existence of a third EAG susceptibility gene (designated Gasa3) on Chr 6. Epistatic interactions between the Gasa2 EAG susceptibility gene and the H2 were also identified, and the presence of an H2-linked susceptibility gene (Gasa4) confirmed by analysis of H2 congenic mice.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Gastritis/genética , Gastritis/inmunología , Animales , Cruzamiento , Mapeo Cromosómico , Genes Dominantes , Genes Recesivos , Ligamiento Genético , Antígenos H-2/genética , Inmunogenética , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
8.
Immunol Today ; 21(11): 546-50, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11094257

RESUMEN

Novel antigen delivery systems are currently being developed by genetic manipulation of the MHC class II trafficking pathway. Specific targeting of endogenously synthesized antigens to the class II loading compartment can result in massively enhanced presentation of peptide epitopes. This emerging technology holds promise for a variety of clinical applications including vaccine development, cancer therapies and control of autoimmune diseases.


Asunto(s)
Presentación de Antígeno , Antígenos de Histocompatibilidad Clase II/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Autoinmunidad , Sitios de Unión , Epítopos/metabolismo , Humanos , Chaperonas Moleculares/inmunología , Vacunas de ADN/inmunología
9.
Int Immunol ; 12(3): 343-52, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10700469

RESUMEN

The catalytic alpha and glycoprotein beta subunits of the gastric H/K ATPase are major molecular targets in human and mouse autoimmune gastritis. We have previously shown that the H/K ATPase beta subunit is required for the initiation of mouse gastritis and identified a gastritogenic H/K ATPase beta subunit peptide (H/Kbeta253-277). Here we report the generation of MHC class II-restricted TCR transgenic mice using V(alpha)9 and V(beta)8.3 TCR chains with specificity for the gastritogenic H/Kbeta253-277 peptide. We found an 8-fold reduction in CD4(+) T cells in the thymus of the transgenic mice. Despite the reduction in intrathymic CD4(+) T cells, V(beta)8. 3-expressing T cells comprised the majority (>90%) of peripheral spleen and lymph node T cells. These peripheral T cells retained their capacity to proliferate in vitro to the H/Kbeta253-277 peptide. Using the responsive T cells, we have restricted the gastritogenic T cell epitope to H/Kbeta261-274. Despite the capacity of the peripheral T cells to proliferate in vitro to the peptide, the majority ( approximately 80%, 13 of 16) of transgenic mice remained free of gastritis while a minority (20%, three of 16) spontaneously developed an invasive and destructive gastritis. Our results confirm that H/Kbeta261-274 is a gastritogenic peptide. The data also suggest that CD4 T cell tolerance to the gastritogenic peptide in the transgenic mice is maintained by a combination of intrathymic and peripheral tolerance mechanisms.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/fisiología , Gastritis/inmunología , ATPasa Intercambiadora de Hidrógeno-Potásio/inmunología , Tolerancia Inmunológica/fisiología , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timo/inmunología , Animales , Autoanticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Mucosa Gástrica/inmunología , Mucosa Gástrica/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/química , Factor Intrinseco/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Parietales Gástricas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Bazo/inmunología , Bazo/patología , Porcinos , Timo/patología
10.
J Leukoc Biol ; 67(2): 169-73, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10670576

RESUMEN

The pathogenesis of autoimmune gastritis is the result of lymphocyte infiltration of the gastric mucosa, however, the events leading to the selective extravasation of autoreactive lymphocytes are unclear. Here we have examined the expression of adhesion molecules in the gastric mucosa of BALB/c mice with neonatal thymectomy-induced gastritis. The overall area of vascular endothelium was not significantly different between gastritic and non-gastritic mice. However, a significant increase in the area of mucosal endothelium expressing MAdCAM-1 in gastritic mice was observed. Treatment of neonatally thymectomized BALB/c mice with a MAdCAM-1 specific monoclonal antibody (MECA 367) reduced the incidence of autoimmune gastritis from 80 to 26%. Treatment with a monoclonal antibody (R1-2) directed to the MAdCAM-1 ligand, alpha4beta7, also resulted in a reduction in the incidence of gastritis to 40%. These findings identify the alpha4beta7/MAdCAM-I interaction as a pivotal event in the initiation of autoimmune gastritis.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Gastritis/metabolismo , Inmunoglobulinas/metabolismo , Mucoproteínas/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/metabolismo , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Linfocitos T CD4-Positivos/inmunología , Moléculas de Adhesión Celular/metabolismo , Endotelio Vascular/metabolismo , Mucosa Gástrica/metabolismo , Gastritis/etiología , Gastritis/inmunología , Gastritis/prevención & control , Inmunoglobulina G/uso terapéutico , Inmunoglobulinas/inmunología , Integrina alfa4 , Ratones , Ratones Endogámicos BALB C , Mucoproteínas/inmunología , Unión Proteica , Ratas , Timectomía/efectos adversos
11.
Am J Physiol Gastrointest Liver Physiol ; 278(2): G266-72, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10666051

RESUMEN

Nonsteroidal anti-inflammatory drug (NSAID) use reduces the risk of colorectal cancer by 40-50%. Previous studies suggest that effective inhibition of colorectal cancer by NSAIDs may be dependent on the presence or absence of a K-ras mutation. This study was aimed at determining the relationship between inhibition of colorectal cancer by sulindac and sulindac sulfone and the presence of activating K-ras mutations in the 1,2-dimethylhydrazine dihydrochloride rat model. Sulindac (20 mg x kg(-1) x day(-1)), sulindac sulfone (40 mg x kg(-1) x day(-1)), or vehicle was administered orally to male Sprague-Dawley rats for a 4-wk period beginning 20 wk after tumor induction. Tumor number and volume were measured before treatment by laparotomy and colonoscopy and again after treatment. Sulindac and sulindac sulfone treatment significantly reduced the number and volume of colorectal tumors compared with control rats. For K-ras (codon 12) mutation detection, frozen tumor tissue was collected at the endpoint. We found K-ras codon 12 mutations in 11 of 21 (52%) control tumors. The proportion of tumors with K-ras mutations in the sulindac-treated group [5 of 8 (62%); odds ratio = 1.51 (95% confidence interval = 0.29, 8.33)] and the proportion of sulindac sulfone-treated tumors [9 of 14 (64%); odds ratio = 1.63 (95% confidence interval = 0.41, 6.66)] were not significantly different from controls. Tumor inhibition did not correlate with K-ras (codon 12) mutation status, which suggests that the mechanism of inhibition of rat colorectal cancer by sulindac and sulindac sulfone is independent of K-ras mutation.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Genes ras , Mutación , Sulindac/análogos & derivados , Sulindac/uso terapéutico , 1,2-Dimetilhidrazina , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinógenos , Codón , Neoplasias del Colon/inducido químicamente , Femenino , Masculino , Ratas , Ratas Sprague-Dawley
12.
Autoimmunity ; 33(1): 1-14, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11204248

RESUMEN

We have previously shown that autoimmune gastritis can be elicited in mice by immunisation with the gastric parietal cell H/K ATPase alphabeta heterodimer, and, furthermore, have identified the H/K ATPase beta-subunit epitope, H/Kbeta253-277 as the dominant epitope of the gastric H/K ATPase. Using gastric H/K ATPase-immunised mice, here we have generated two T cell hybridomas specific for the H/Kbeta253-277 peptide, namely 4B11.F4.5 and 1E4.C1. Hybridoma 4B11.F4.5 uses Valpha8 and Vbeta8.2 TCR chains and 1E4.C1 uses Valpha9 and V1beta8.3 chains. Although both hybridomas are specific for H/Kbeta253-277, T cell assays using overlapping 14-mers of the 25-mer epitope showed that the two autoreactive TCRs recognise different regions of the 25-mer. The TCR from 1E4.C1 has been used to generate a TCR beta-chain transgenic mouse. >80% of peripheral CD4+ T cells utilise the Vbeta8.3 transgene. As expected, 1E4-TCR beta-chain transgenic mice are susceptible to neonatal thymectomy induced autoimmune gastritis. While none of the 1E4-TCR beta chain transgenic mice spontaneously developed a destructive gastritis, a minority (20%) of the transgenic mice developed a non-invasive and non-destructive gastritis. This suggests that the pathogenic T cells are maintained in a tolerant state in the periphery of the transgenic mice.


Asunto(s)
Autoantígenos/inmunología , Epítopos de Linfocito T/inmunología , ATPasa Intercambiadora de Hidrógeno-Potásio/inmunología , Epítopos Inmunodominantes/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Estómago/enzimología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Epitopo , Femenino , Hibridomas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Estómago/inmunología
13.
Eur J Immunol ; 29(11): 3768-81, 1999 11.
Artículo en Inglés | MEDLINE | ID: mdl-10556834

RESUMEN

NK1.1(+)alpha betaTCR(+) (NKT) cells have several important roles including tumor rejection and prevention of autoimmune disease. Although both CD4(+) and CD4(-)CD8(-) double-negative (DN) subsets of NKT cells have been identified, they are usually described as one population. Here, we show that NKT cells are phenotypically, functionally and developmentally heterogeneous, and that three distinct subsets (CD4(+), DN and CD8(+)) are differentially distributed in a tissue-specific fashion. CD8(+) NKT cells are present in all tissues but the thymus, and are highly enriched for CD8alpha(+)beta(-) cells. These subsets differ in their expression of a range of cell surface molecules (Vbeta8, DX5, CD69, CD45RB, Ly6C) and in their ability to produce IL-4 and IFN-gamma, with splenic NKT cell subsets producing lower levels than thymic NKT cells. Developmentally, most CD4(+) and DN NKT cells are thymus dependent, in contrast to CD8(+) NKT cells, and are also present amongst recent thymic emigrants in spleen and liver. TCR Jalpha281-deficient mice show a dramatic deficiency in thymic NKT cells, whereas a significant NKT cell population (enriched for the DN and CD8(+) subsets) is still present in the periphery. Taken together, this study reveals a far greater level of complexity within the NKT cell population than previously recognized.


Asunto(s)
Antígenos/inmunología , Células Asesinas Naturales/inmunología , Proteínas/inmunología , Animales , Antígenos Ly , Antígenos de Superficie , Antígenos CD4/inmunología , Movimiento Celular , Supervivencia Celular , Femenino , Inmunofenotipificación , Interferón gamma/biosíntesis , Interleucina-4/biosíntesis , Células Asesinas Naturales/clasificación , Lectinas Tipo C , Ratones , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timo/inmunología
14.
Gastroenterology ; 117(3): 605-18, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10464136

RESUMEN

BACKGROUND & AIMS: Parietal cells of the gastric mucosa contain a complex and extensive secretory membrane system that harbors gastric H(+),K(+)-adenosine triphosphatase (ATPase), the enzyme primarily responsible for acidification of the gastric lumen. We have produced mice deficient in the H(+),K(+)-ATPase beta subunit to determine the role of the protein in the biosynthesis of this membrane system and the biology of gastric mucosa. METHODS: Mice deficient in the H(+), K(+)-ATPase beta subunit were produced by gene targeting. RESULTS: The stomachs of H(+),K(+)-ATPase beta subunit-deficient mice were achlorhydric. Histological and immunocytochemical analyses with antibodies to the H(+),K(+)-ATPase alpha subunit revealed that parietal cell development during ontogeny was retarded in H(+), K(+)-ATPase beta subunit-deficient mice. In 15-day-old mice, cells with secretory canaliculi were observed in wild-type but not in H(+), K(+)-ATPase beta subunit-deficient mice. Parietal cells of H(+), K(+)-ATPase beta subunit-deficient mice 17 days and older contained an abnormal canaliculus that was dilated and contained fewer and shorter microvilli than normal. In older parietal cells, the abnormal canaliculus was massive (25 micrometer in diameter) and contained few microvilli. We did not observe typical tubulovesicular membranes in any parietal cell from H(+),K(+)-ATPase beta subunit-deficient mice. Histopathologic alterations were only observed in the stomach. CONCLUSIONS: The H(+),K(+)-ATPase beta subunit is required for acid-secretory activity of parietal cells in vivo, normal development and cellular homeostasis of the gastric mucosa, and attainment of the normal structure of the secretory membranes.


Asunto(s)
Mucosa Gástrica/citología , ATPasa Intercambiadora de Hidrógeno-Potásio/fisiología , Células Parietales Gástricas/fisiología , Animales , Mucosa Gástrica/enzimología , Mucosa Gástrica/metabolismo , Gastrinas/sangre , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Concentración de Iones de Hidrógeno , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/ultraestructura , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Am J Physiol ; 277(1): G209-18, 1999 07.
Artículo en Inglés | MEDLINE | ID: mdl-10409169

RESUMEN

We have investigated the underlying basis of the lesion in murine autoimmune gastritis, a model of the human disease pernicious anemia. The disease is mediated by T lymphocytes and characterized by selective depletion of parietal and zymogenic cells from the gastric unit (gland) together with gastric epithelial cell hyperplasia. The gastric units of gastritic stomachs contained 2.3-fold more cells than normal and accumulated rapidly dividing, short-lived gastric epithelial stem cells and mucous neck cells. Most of these immature cells failed to differentiate into end-stage cells but rather appeared to die by apoptosis. We also found no correlation between anti-parietal cell autoantibody titers and the degree of gastric pathology, providing further evidence that autoantibodies do not play a direct role in the pathogenesis of gastritis. Taken together, the normal developmental pathways of the gastric mucosa are disrupted in autoimmune gastritis, resulting in an amplification of immature cell types. The differentiation of these immature cells appears to be blocked, contributing to depletion of end-stage cells. This scenario provides an explanation for depletion of not only parietal cells but also zymogenic cells even though they are not directly targeted by the immune system.


Asunto(s)
Enfermedades Autoinmunes/patología , Mucosa Gástrica/patología , Gastritis/patología , Animales , Autoanticuerpos/análisis , Enfermedades Autoinmunes/inmunología , Muerte Celular/fisiología , División Celular/fisiología , Mucosa Gástrica/inmunología , Gastritis/inmunología , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Fenotipo , Timectomía , Factores de Tiempo
16.
Glycobiology ; 9(6): 601-6, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10336993

RESUMEN

The gastric H/K ATPase beta-subunit, an abundant glycoprotein of the secretory membranes of gastric parietal cells, is the major autoantigen recognized by human parietal cell autoantibodies in gastric autoimmunity. Our previous studies demonstrated that the human autoantibodies recognize the H/K ATPase beta-subunit from a number of species and that glycosylation of the beta-subunit with complex N-glycans is required for autoantibody binding. The N-glycans of the beta-subunit contain polylactosamine chains. The lactosamine chains of the rabbit beta-subunit are terminated with alpha-linked galactosyl residues (alpha-galactosyl epitope) (Tyagarajan et al., Biochemistry, 1996, 35, 3238-3246). Here we have investigated the expression of alpha-galactosyl epitopes on the H/K ATPase beta-subunit from a number of species. Using the alpha-galactosyl binding lectin, BS1-IB4, and naturally occurring anti-alpha-galactosyl antibodies, we have demonstrated that the rat H/K ATPase beta-subunit also contains terminal alpha-galactosyl residues, but not the beta-subunit from pig, dog, and mouse, indicating species-specific differences in the terminal saccharide sequences of the beta-subunit. We also investigated the potential contribution of the alpha-galactosyl epitopes to the binding by human sera. The reactivity of human pernicious anemia serum with gastric parietal cells could not be inhibited with saccharide inhibitors and, in addition, no binding was observed with normal human sera. We conclude that the H/K ATPase beta-subunit oligosaccharides from rabbit and rat are terminated with alpha-galactosyl epitopes, and although the presence of this epitope does not contribute to binding by human parietal cell autoantibodies at the concentrations routinely used, it is recommended that neither rat or rabbit stomachs be used for screening human sera.


Asunto(s)
Autoanticuerpos/inmunología , Epítopos/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Células Parietales Gástricas/inmunología , Estómago/enzimología , Animales , Galactosa/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/inmunología , Humanos , Especificidad de la Especie , Estómago/inmunología
17.
Immunology ; 96(1): 145-51, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10233689

RESUMEN

We have previously shown that autoimmune gastritis can be elicited in mice by immunization with the gastric parietal cell H/K ATPase alpha beta heterodimer and that tolerance specifically induced to the H/K ATPase beta-subunit protects mice from the development of gastritis. Here we have identified the immunodominant gastritogenic epitope of the H/K ATPase beta-subunit (H/Kbeta). Epitope mapping was carried out with a panel of 21 overlapping peptides that spanned the entire sequence of the gastric H/K ATPase beta-subunit. T cells from gastric H/K ATPase-immunized mice responded to only one of the overlapping peptides, namely H/Kbeta253-277. Furthermore, a single subcutaneous immunization of 6-week-old BALB/c mice with the ATPase beta-subunit peptides resulted in a T-cell response to only H/Kbeta253-277. Multiple immunization with the overlapping H/K ATPase peptides demonstrated that H/Kbeta253-277 was capable of inducing a mononuclear infiltrate specifically within the gastric mucosa. We conclude that H/Kbeta253-277 is the dominant gastritogenic epitope of the gastric H/K ATPase.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Epítopos/análisis , Mucosa Gástrica/inmunología , Gastritis/inmunología , ATPasa Intercambiadora de Hidrógeno-Potásio/inmunología , Isoenzimas/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Enfermedades Autoinmunes/enzimología , Mapeo Epitopo , Mucosa Gástrica/enzimología , Gastritis/enzimología , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Isoenzimas/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular
18.
J Immunol ; 162(9): 5106-11, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10227980

RESUMEN

Although much is known about the pathology of human chronic atrophic (type A, autoimmune) gastritis, its cause is poorly understood. Mouse experimental autoimmune gastritis (EAG) is a CD4+ T cell-mediated organ-specific autoimmune disease of the stomach that is induced by neonatal thymectomy of BALB/c mice. It has many features similar to human autoimmune gastritis. To obtain a greater understanding of the genetic components predisposing to autoimmune gastritis, a linkage analysis study was performed on (BALB/cCrSlc x C57BL/6)F2 intercross mice using 126 microsatellite markers covering 95% of the autosomal genome. Two regions with linkage to EAG were identified on distal chromosome 4 and were designated Gasa1 and Gasa2. The Gasa1 gene maps within the same chromosomal segment as the type 1 diabetes and systemic lupus erythematosus susceptibility genes Idd11 and Nba1, respectively. Gasa2 is the more telomeric of the two genes and was mapped within the same chromosomal segment as the type 1 diabetes susceptibility gene Idd9. In addition, there was evidence of quantitative trait locus controlling autoantibody titer within the telomeric segment of chromosome 4. The clustering of genes conferring susceptibility to EAG with those conferring susceptibility to type 1 diabetes is consistent with the coinheritance of gastritis and diabetes within human families. This is the first linkage analysis study of autoimmune gastritis in any organism and as such makes an important and novel contribution to our understanding of the etiology of this disease.


Asunto(s)
Enfermedades Autoinmunes/genética , Cromosomas/inmunología , Gastritis/genética , Gastritis/inmunología , Ligamiento Genético/inmunología , Predisposición Genética a la Enfermedad/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Enfermedades Autoinmunes/inmunología , Mapeo Cromosómico , Cruzamientos Genéticos , Femenino , Marcadores Genéticos , Inmunidad Innata/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Carácter Cuantitativo Heredable , Programas Informáticos
19.
Int Immunol ; 10(10): 1491-9, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9796916

RESUMEN

Neonatal thymectomy of mice 3 days after birth but not at birth leads to T cell-mediated, organ-specific, autoimmune disease in a strain-dependent manner. The mechanisms that lead to disease in this model remain unknown, but the answer may lie in a deficiency of thymus-dependent cells or factors. One candidate is the relatively rare population of NK1.1 + T cells (NKT cells). Conventional alphabetaTCR+ T cells appear in the thymus from days 17-18 of embryogenesis and start emigrating to the periphery around birth, whereas the development of NKT cells is thought to be delayed until at least 1 week after birth. We have confirmed this to be the case in both (BALB/c x C57BL/6)F1 (autoimmune susceptible) and C57BL/6 (autoimmune resistant) mice. Moreover, examination of T cells (in spleen, lymph nodes, liver and bone marrow) from mice following 3 day neonatal thymectomy revealed a significant reduction in the presence of NKT cells in all tissues. However, the extent of depletion was generally more pronounced in (BALB/c x C57BL/6)F1 than in C57BL/6 mice, and the few remaining NKT cells in C57BL/6 mice were enriched for a CD4-CD8int subset which is absent from the thymus and may represent a distinct lineage of thymus-independent NKT cells. Given mounting evidence of a role for NKT cells in protection from autoimmune disease, it is possible that their specific removal by neonatal thymectomy may contribute to the susceptibility of these mice to autoimmune disease.


Asunto(s)
Linfocitos T/citología , Timectomía , Animales , Animales Recién Nacidos , Complejo CD3/análisis , Antígenos CD4/análisis , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Timo/fisiología
20.
Immunology ; 93(3): 405-8, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9640252

RESUMEN

Murine autoimmune gastritis, induced by neonatal thymectomy, is characterized by a mononuclear infiltrate within the gastric mucosa, loss of parietal and zymogenic cells and circulating autoantibodies to the gastric H/K ATPase. The infiltrate contains both CD4+ and CD8+ T cells. Here we have investigated the roles of CD4+ and CD8+ T cells in the development of gastritis by in vivo treatment with depleting rat anti-CD4 and anti-CD8 monoclonal antibodies. Depletion of CD4+ T cells decreased the incidence of gastric mononuclear infiltrates from 63% (5/8), observed in normal rat immunoglobulin G (IgG)-injected mice, to 8% (1/12) and also abolished the production of antigastric autoantibodies. In contrast, depletion of CD8+ T cells did not reduce the incidence of gastritis. The absence of CD8+ T cells in the infiltrate of the stomach of anti-CD8(+)-treated mice was confirmed by immunocytochemistry. These results argue that neonatal thymectomy-induced autoimmune gastritis is mediated by CD4+ T cells and that CD8+ T cells do not play a significant role in the development of the gastric lesion.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Gastritis/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Inmunoglobulina G/inmunología , Inmunohistoquímica , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Timectomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA